JGV Papers in Press. Published March 11, 2015 as doi:10.1099/vir.0.000120

Journal of General Virology CD4 binding site broadly neutralizing antibody selection of HIV-1 escape mutants --Manuscript Draft-Manuscript Number:

VIR-D-15-00017R2

Full Title:

CD4 binding site broadly neutralizing antibody selection of HIV-1 escape mutants

Short Title:

selection of HIV-1 neutralization escape mutants

Article Type:

Short Communication

Section/Category:

Animal - Retroviruses

Corresponding Author:

Aine McKnight Queen Mary's School of Medicine and Dentistry London, UNITED KINGDOM

First Author:

Hanna Dreja

Order of Authors:

Hanna Dreja Corinna Pade Lei Chen Aine McKnight

Abstract:

All human-immunodeficiency-virus type-1 (HIV-1) viruses use CD4 to enter cells. Consequently the viral envelope CD4-binding-site (CD4bs) is relatively conserved, making it a logical neutralizing antibody target. It is important to understand how CD4binding site variation allows for escape from neutralizing antibodies. Alanine scanning mutagenesis identifies residues in antigenic sites, whereas escape mutant selectionI dentifies viable mutants. We selected HIV-1 to escape CD4bs neutralizing MAbs b12, A12 and HJ16. Viruses that escape from A12- and b12- remained susceptible to HJ16, VRC01 and J3, whilst six different viruses that escape HJ16 remained sensitive to A12, b12 and J3. In contrast, their sensitivity to VRC01 was variable. Triple HJ16/A12/b12resistant virus proved that HIV-1 can escape multiple BNMAbs, but still retain sensitivity to VRC01 and the llama derived J3 nanobody. This antigenic variability could reflect that occurring in circulating viruses so studies like this could predict immunologically relevant antigenic forms of the CD4bs for inclusion in HIV-1 vaccines.

Powered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporation

Manuscript Including References (Word document) Click here to download Manuscript Including References (Word document): clean MS VIR-D-15-00017R2.docx

1 2 3

CD4 binding site broadly neutralizing antibody selection of HIV-1 escape mutants

4

Hanna Dreja1, Corinna Pade1, Lei Chen2, Áine McKnight*1

5

1

6 7

Centre for Immunology and Infectious Disease, Blizard Institute, Barts & The London School of Medicine and Dentistry, Queen Mary University of London 2

Vaccine Research Center, NIAID, National Institutes of Health, Bethesda, Maryland

8 9

Running Title: Neutralizing antibody selection of HIV-1 escape mutants

10 11

*Corresponding Author: Áine McKnight email [email protected]

12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28

Words 2229

29

Abstract

30

All human-immunodeficiency-virus type-1 (HIV-1) viruses use CD4 to enter cells. Consequently the

31

viral envelope CD4-binding-site (CD4bs) is relatively conserved, making it a logical neutralizing

32

antibody target. It is important to understand how CD4-binding site variation allows for escape

33

from neutralizing antibodies. Alanine scanning mutagenesis identifies residues in antigenic sites,

34

whereas escape mutant selection identifies viable mutants. We selected HIV-1 to escape CD4bs

35

neutralizing MAbs b12, A12 and HJ16. Viruses that escape from A12- and b12- remained

36

susceptible to HJ16, VRC01 and J3, whilst six different viruses that escape HJ16 remained sensitive

37

to A12, b12 and J3. In contrast, their sensitivity to VRC01 was variable. Triple HJ16/A12/b12-

38

resistant virus proved that HIV-1 can escape multiple BNMAbs, but still retain sensitivity to VRC01

39

and the llama derived J3 nanobody. This antigenic variability could reflect that occurring in

40

circulating viruses so studies like this could predict immunologically relevant antigenic forms of the

41

CD4bs for inclusion in HIV-1 vaccines.

42 43 44 45 46 47 48 49 50 51 52 53 54 55 56

57

A successful human immunodeficiency virus type 1 (HIV-1) vaccine is expected to need to induce

58

robust CD4+ and CD8+ cellular responses, in concert with a strong and broadly neutralizing

59

antibody response. Designing immunogens that trigger such responses is challenging (reviewed in

60

(Haynes & Montefiori, 2006; McCoy & Weiss, 2013)), partly due to the diversity (Gaschen et al.,

61

2002) of the viral envelope glycoprotein (Env), which interacts with cell receptors including CD4.

62

Hence, the CD4 binding site (CD4bs) is functionally conserved and is therefore a logical vaccine

63

target to elicit neutralizing Abs. A number of highly effective anti-CD4bs broadly neutralizing

64

monoclonal antibodies (BNMAbs) with distinct neutralization profiles have been generated from

65

HIV-1 infected individuals (Burton et al., 1991; Burton et al., 1994; Corti et al., 2010; Falkowska et

66

al., 2012; Walker et al., 2009; Wu et al., 2010; Zhou et al., 2010) or vaccinated llamas (Forsman et

67

al., 2008; McCoy et al., 2012). A full understanding of the nature of these antibodies would

68

provide clues to the antigenic landscape of the CD4bs, which may be important for developing an

69

inclusive HIV-1 vaccine. In addition to structurally define current BNMAbs and their corresponding

70

CD4bs footprints, the ability of replication competent viruses to escape such antibodies and to

71

determine whether the mutants remain susceptible to alternative anti-CD4bs BNMAbs will be

72

valuable.

73

We used the three different anti-CD4bs BNMAbs b12, A12 and HJ16 to select for escape mutants.

74

B12 was the first human BNMAb to map to the CD4bs and competes for binding to soluble CD4

75

(sCD4) (Barbas et al., 1992; Burton et al., 1991; Burton et al., 1994; Roben et al., 1994; Zhou et al.,

76

2010). The llama-derived single chain Ab A12 also competes with b12 and sCD4 (Forsman et al.,

77

2008), as does HJ16 (Corti et al., 2010). Alanine scanning has determined that HJ16 belongs to a

78

class of antibodies, which binds to a region distinct from the classic CD4bs (AA 474-476) (Pietzsch

79

et al., 2010). Recently, Balla-Jhagjhoorsingh et al (Balla-Jhagjhoorsingh et al., 2013) identified a

80

glycosylation site (N276) critical for HJ16-induced escape of a primary HIV-1 strain in an in vivo

81

model.

82

We used the well described HIV-1 replication competent clone HXB2 (Ratner et al., 1985) as it is

83

unlike primary isolates highly sensitive to many anti-CD4bs BNMAbs. This is probably because

84

HXB2 was highly passaged in vitro in the absence of humoral responses Escape viruses were

85

selected in C8166 CD4+ T-cells (Salahuddin et al., 1983) in gradually increasing concentrations of

86

BNMAbs (from 50 ng NMAb ml-1). Cell free supernatants were harvested from cells with cytopathic

87

appearances and added to target cells for a second round of infection, this time with a doubling of

88

the BNMAb concentration. After two to four weeks, resistant viruses emerged that could replicate

89

in the presence of high concentrations of each BNMAb (10 µg A12 or HJ16 ml–1 and 20 µg b12 ml–

90

1

91

resistant viruses were PCR amplified and sequenced (Dreja et al., 2010).

92

Selection of HXB2 with b12 resulted in a virus with the single dominant AA change (G366E) within

93

the envelope CD4bs (Fig 1, 3). This change is located three AAs upstream of the proline to lysine

94

mutation seen in a previous escape study (Mo et al, 1997), where additionally two mutations were

95

observed in the V2 region. Interestingly, we did not identify any additional changes. This lack of

96

other mutations within the V2 could be explained by the ease of neutralisation of HXB2,

97

suggesting that supplementary V2 compensatory mutations were not required for resistance. It

98

may be that the G366E mutation did not hamper viral infectivity. Glycine 366 has previously been

99

implicated in b12 binding (Li et al., 2011; Saphire et al., 2001, Zhou et al., 2007). The site is

100

mapped to a model of the crystal structure of the trimeric HIV-1 Env spike, (Fig 3). The HIV-1 viral

101

env-expression vector psvIII-HXB2 (Gao et al., 1996) was engineered by site-directed mutagenesis

102

(SDM) to carry the G366E mutation. SDM-pseudotyped virions carrying the luciferase reporter

103

gene were produced as previously described (Dreja et al., 2010). The resulting SDM(b12)

104

confirmed the b12 resistant phenotype (Fig 2a). The mutation had no effect on viral susceptibility

105

to A12 and HJ16, even though both these BNMAbs compete for binding with b12, with each other

106

and with sCD4. Moreover the sensitivity of SDM(b12) to VRC01 (Wu et al., 2010; Zhou et al., 2010)

107

remained the same (Fig 2b). The recently described llama derived anti-CD4 BNMAb J3 (McCoy et

108

al., 2012) neutralized SDM(b12) as efficiently as wild type (WT)-HXB2 (Fig 2b). These results

109

suggest that the AA change responsible for b12 escape is antigenically distinct from those of the

110

human BNMAbs HJ16 and VRC01 and the llama BNMAbs A12 and J3. The viral escape from b12

111

had little effect on the ability of virus to replicate in vitro (data not shown) but was measurably

112

less sensitive to CD4-IgG2 inhibition (IC50 shifted from 5ng ml-1 to 50 ng ml-1 and for the

113

pseudotype SDM(b12) from 10ng ml-1 to 100 ng ml-1, Fig 2b).

114

The A12-selected escape virus was resistant to A12 and carried the S375N mutation adjacent to

115

the CD4bs (Fig 1 and 3). SDM(A12) confirmed that A12 resistance is conferred entirely by this

116

mutation (Fig 2a). SDM(A12) remained sensitive to both b12 and HJ16, supporting the notion that

117

the epitopes of A12 and b12 or HJ16 are distinct. The J3 llama antibody neutralized SDM(A12) and

118

there is a small reduction in sensitivity to VRC01 for SDM(A12). There was, however, no apparent

119

effect on the sensitivity of the mutation in SDM(A12) to CD4-IgG2.

). Neutralization assays were carried out and proviral full length env from cells infected with

120

In contrast to the b12 and A12 selection, where dominant genotypes were generated, selection

121

with HJ16 yielded a viral swarm, containing several different mutant viruses. It could be that there

122

are more options for escape routes with this single chain antibody compared to bivalent

123

antibodies which may have more steric hindrance. The proviral env sequences amplified from HJ16

124

viral selected cultures (from nucleotide 127 (KpnI) to 2251 (BamHI)) were inserted into the env-

125

expression vector psvIII-HXB2 and six different infectious, resistant clones were identified.

126

Interestingly, and in keeping with the observation that glycosylation may be associated with HJ16

127

resistance in primary cell cultures (Balla-Jhagjhoorsingh et al., 2013), we also observed that four

128

mutations of a potential N-linked glycosylation site in the V5 loop affected sensitivity to HJ16

129

neutralization (N463S, S465F, S465P and S465Y) (Fig 1, 3). Gray et al, (Gray et al., 2011)

130

demonstrated a relatively high degree of sequence variation within the V5 loop in a large,

131

independent panel of Envs, which may affect the accessibility to the CD4bs. Remarkably, none of

132

these four substitutions significantly affected CD4-IgG2 inhibition (Fig 2b). The mutations are

133

eleven and nine AAs upstream of the core region (474-476), identified as a HJ16 target by Pietzsch

134

et al (Pietzsch et al., 2010). Curiously, of the three HJ16 resistant pseudoviruses with substitutions

135

at position 465, two (psHJ16(S465F) and psHJ16(S465P)) gained sensitivity to VCR01 (Fig 2a). This

136

is concurrent with alanine substitution of this residue (Falkowska et al., 2012), which increased

137

sensitivity to VRC01 neutralization. In contrast, psHJ16(S456Y) retained wildtype sensitivity to

138

VRC01. This suggests that the glycosylation per se is not important for the antibody footprint of

139

VRC01, although it appears crucial for HJ16 activity. Similarly, psHJ16(N463S) maintained wildtype

140

sensitive to VRC01. Overall our results suggest that the V5 region is involved in HJ16 and VRC01

141

binding, as changes in this domain affect neutralization to both BNMAbs. The fifth HJ16 resistant

142

virus had a glycine to aspartic acid change at position 459 (psHJ16D(G459D)), and resulted in a

143

virus that was marginally more resistant to VRC01. This mutation was identified in HIV-1 (JRCSF)

144

infected humanised mice treated with 45-46G54W, a BNMAb belonging to the VRC01 family (Klein

145

et al, 2012). The G459D mutation is only four AAs upstream of the glycosylation site in the V5 loop,

146

and exhibits a similar neutralisation profile as psHJ16(N463S) . By contrast, similar to S465F and

147

S465P, the HJ16 resistant clone E409R also became more neutralization sensitive to VRC01 (Fig

148

2b). All HJ16 resistant pseudotyped viruses retained sensitivity to J3, b12, CD4-IgG2 and, in four

149

cases, to A12. Interestingly, G459D and E409R appeared more sensitive to A12 neutralisation at

150

lower concentrations (1000 ng x mL-1). This partial VRC01 resistance is in accordance with the findings from

176

the individual AA changes, where the N463S and G366E mutations were neutral, whereas S375N

177

renders the virus less sensitive to VRC01 neutralization (Fig 2b). SDM(HJ16/A12/b12) was

178

marginally less sensitive to J3 (IC50 from 3 to 25 ng x mL-1), as predicted by the lower sensitivity of

179

the S375N and G366E mutations (Fig 2b). As expected, the sensitivity to CD4-IgG was partly lost,

180

which is likely to be due to the G366E mutation described above (IC50 from 2 to 20 ng x mL-1).

181

Nevertheless, it is intriguing that albeit marginally resistant to the BNMAbs VRC01 and J3, the

182

triply selected virus retains some sensitivity to these BNMAbs. This suggests that if one succeeds in

183

inducing a range of different anti-CD4bs Abs by vaccination, neutralization control of HIV-1 may be

184

achieved. Also, the three BNMAbs, provided at the same time, never enabled for a resistant virus

185

to emerge, cautiously suggesting that there is a limit to the amount of pressure the virus can

186

withstand. Pre-exposure prophylaxis using modified BNMAb is considered as an option to prevent

187

HIV-1 acquisition (Pace et al., 2013). Our findings would support such an approach and suggest

188

that multiple BNMAbs targeting the CD4bs structure should be considered.

189

Studies of the antigenic landscape of BNMAbs and the escape routes viruses master can help us to

190

target important immunogenic epitopes for HIV-1 vaccines, but also to consider including

191

anticipated escape structures.

192 193

Authors' contributions

194

HD and AM conceived and designed the experiments. HD and CP acquired the data. HD and AM

195

interpreted the data and drafted the manuscript. LC mapped mutations onto HIV-1 trimer and

196

provided structural context to data. All authors read and approved the final manuscript.

197

Acknowledgements

198

B12 was provided by Dennis Burton, The Scripps Research Institute; A12 and J3 from Robin Weiss,

199

UCL; HJ16 from Humabs Biomed SA, Switzerland. VRC01 was obtained through the AIDS Research

200

and Reference Reagent Program, Division of AIDS, NIAID, NIH, from John Mascola and CD4-IgG2

201

through the NIH AIDS Reagent Program, Division of AIDS, NIAID, NIH: Cat#11780 from Progenics

202

Pharmaceuticals. The genetic replication competent clone HXB2 was obtained from the Centre for

203

AIDS Research (NIBSC, UK). psvIII-HXB2 was provided by Paul Clapham, Worcester, MA. This

204

research was conducted as part of the Collaboration for AIDS Vaccine Discovery funded by the Bill

205

and Melinda Gates Foundation (Weiss VDAC, UCL). We would like to thank Robin Weiss, Peter

206

Kwong and Laura McCoy for critically reading the manuscript.

207

References

208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258

Balla-Jhagjhoorsingh, S. S., Corti, D., Heyndrickx, L., Willems, E., Vereecken, K., Davis, D. & Vanham, G. (2013). The N276 glycosylation site is required for HIV-1 neutralization by the CD4 binding site specific HJ16 monoclonal antibody. PLoS One 8, e68863. Barbas, C. F., 3rd, Bjorling, E., Chiodi, F., Dunlop, N., Cababa, D., Jones, T. M., Zebedee, S. L., Persson, M. A., Nara, P. L. & other authors (1992). Recombinant human Fab fragments neutralize human type 1 immunodeficiency virus in vitro. Proc Natl Acad Sci U S A 89, 9339-9343. Burton, D. R., Barbas, C. F., 3rd, Persson, M. A., Koenig, S., Chanock, R. M. & Lerner, R. A. (1991). A large array of human monoclonal antibodies to type 1 human immunodeficiency virus from combinatorial libraries of asymptomatic seropositive individuals. Proc Natl Acad Sci U S A 88, 10134-10137. Burton, D. R., Pyati, J., Koduri, R., Sharp, S. J., Thornton, G. B., Parren, P. W., Sawyer, L. S., Hendry, R. M., Dunlop, N. & other authors (1994). Efficient neutralization of primary isolates of HIV-1 by a recombinant human monoclonal antibody. Science 266, 1024-1027. Corti, D., Langedijk, J. P., Hinz, A., Seaman, M. S., Vanzetta, F., Fernandez-Rodriguez, B. M., Silacci, C., Pinna, D., Jarrossay, D. & other authors (2010). Analysis of memory B cell responses and isolation of novel monoclonal antibodies with neutralizing breadth from HIV-1-infected individuals. PLoS One 5, e8805. Dreja, H., O'Sullivan, E., Pade, C., Greene, K. M., Gao, H., Aubin, K., Hand, J., Isaksen, A., D'Souza, C. & other authors (2010). Neutralization activity in a geographically diverse East London cohort of human immunodeficiency virus type 1-infected patients: clade C infection results in a stronger and broader humoral immune response than clade B infection. J Gen Virol 91, 2794-2803. Falkowska, E., Ramos, A., Feng, Y., Zhou, T., Moquin, S., Walker, L. M., Wu, X., Seaman, M. S., Wrin, T. & other authors (2012). PGV04, an HIV-1 gp120 CD4 binding site antibody, is broad and potent in neutralization but does not induce conformational changes characteristic of CD4. J Virol 86, 43944403. Forsman, A., Beirnaert, E., Aasa-Chapman, M. M., Hoorelbeke, B., Hijazi, K., Koh, W., Tack, V., Szynol, A., Kelly, C. & other authors (2008). Llama antibody fragments with cross-subtype human immunodeficiency virus type 1 (HIV-1)-neutralizing properties and high affinity for HIV-1 gp120. J Virol 82, 12069-12081. Gao, F., Morrison, S. G., Robertson, D. L., Thornton, C. L., Craig, S., Karlsson, G., Sodroski, J., Morgado, M., Galvao-Castro, B. & other authors (1996). Molecular cloning and analysis of functional envelope genes from human immunodeficiency virus type 1 sequence subtypes A through G. The WHO and NIAID Networks for HIV Isolation and Characterization. J Virol 70, 1651-1667. Gaschen, B., Taylor, J., Yusim, K., Foley, B., Gao, F., Lang, D., Novitsky, V., Haynes, B., Hahn, B. H. & other authors (2002). Diversity considerations in HIV-1 vaccine selection. Science 296, 2354-2360. Gray, L., Sterjovski, J., Ramsland, P. A., Churchill, M. J. & Gorry, P. R. (2011). Conformational alterations in the CD4 binding cavity of HIV-1 gp120 influencing gp120-CD4 interactions and fusogenicity of HIV-1 envelopes derived from brain and other tissues. Retrovirology 8, 42. Haynes, B. F. & Montefiori, D. C. (2006). Aiming to induce broadly reactive neutralizing antibody responses with HIV-1 vaccine candidates. Expert Rev Vaccines 5, 347-363. Julien JP, Cupo A, Sok D, Stanfield RL, Lyumkis D, Deller MC, Klasse PJ, Burton DR, Sanders RW, & other authors (2013)Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science, 342, 14771483. Klein, F', Halper-Stromberg A, Horwitz, J A. Gruell H, Scheid, Johannes F. Bournazos, S.Mouquet, H. Spatz, L. A, Diskin R & Others (2012). HIV therapy by a combination of broadly neutralizing antibodies in humanized mice Nature 492,118–12Li, Y., O'Dell, S., Walker, L. M., Wu, X., Guenaga, J., Feng, Y., Schmidt, S. D., McKee, K., Louder, M. K. & other authors (2011). Mechanism of neutralization by the broadly neutralizing HIV-1 monoclonal antibody VRC01. J Virol 85, 8954-8967. McCoy, L. E. & Weiss, R. A. (2013). Neutralizing antibodies to HIV-1 induced by immunization. J Exp Med 210, 209-223.

259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297

McCoy, L. E., Quigley, A. F., Strokappe, N. M., Bulmer-Thomas, B., Seaman, M. S., Mortier, D., Rutten, L., Chander, N., Edwards, C. J. & other authors (2012). Potent and broad neutralization of HIV-1 by a llama antibody elicited by immunization. J Exp Med 209, 1091-1103. Mo, H. Stramatatos, L, Carlos, J. E I.P, Barbas, Parren P.W.H, Burton D.R, Moore, J.P, & Ho, D.D. (1997). Human Immunodeficiency Virus Type 1 Mutants That Escape Neutralization by Human MonoclonalAntibody IgG1b12. J Virol 9 , 6869-6874 Mo, H., Stramatatos, L., Carlos, Ip, J. E, Barbas, C. F., Parren, P. W. H., Burton, D. R., Moore, J. P., & Ho, D.D. (1997). Human Immunodeficiency Virus Type 1 Mutants That Escape Neutralization by Human Monoclonal Antibody IgG1b12. J Virol 9 , 6869-6874 Pace, C. S., Song, R., Ochsenbauer, C., Andrews, C. D., Franco, D., Yu, J., Oren, D. A., Seaman, M. S. & Ho, D. D. (2013). Bispecific antibodies directed to CD4 domain 2 and HIV envelope exhibit exceptional breadth and picomolar potency against HIV-1. Proc Natl Acad Sci U S A 110, 13540-13545. Pancera, M., Majeed, S., Ban, Y.E., Chen, L., Huang, C.C., Kong, L., Kwon, Y.D., Stuckey, J., Zhou, T., Robinson, J.E., Schief, W.R., Sodroski, J., Wyatt, R., Kwong, P.D. (2010). Structure of HIV-1 gp120 with gp41-interactive region reveals layered envelope architecture and basis of conformational mobility. P Proc Natl Acad Sci U S A 107: 1166-1171. Pietzsch, J., Scheid, J. F., Mouquet, H., Klein, F., Seaman, M. S., Jankovic, M., Corti, D., Lanzavecchia, A. & Nussenzweig, M. C. (2010). Human anti-HIV-neutralizing antibodies frequently target a conserved epitope essential for viral fitness. J Exp Med 207, 1995-2002. Ratner, L., Haseltine, W., Patarca, R., Livak, K. J., Starcich, B., Josephs, S. F., Doran, E. R., Rafalski, J. A., Whitehorn, E. A. & other authors (1985). Complete nucleotide sequence of the AIDS virus, HTLV-III. Nature 313, 277-284. Roben, P., Moore, J. P., Thali, M., Sodroski, J., Barbas, C. F., 3rd & Burton, D. R. (1994). Recognition properties of a panel of human recombinant Fab fragments to the CD4 binding site of gp120 that show differing abilities to neutralize human immunodeficiency virus type 1. J Virol 68, 4821-4828. Salahuddin, S. Z., Markham, P. D., Wong-Staal, F., Franchini, G., Kalyanaraman, V. S. & Gallo, R. C. (1983). Restricted expression of human T-cell leukemia--lymphoma virus (HTLV) in transformed human umbilical cord blood lymphocytes. Virology 129, 51-64. Saphire, E. O., Parren, P. W., Pantophlet, R., Zwick, M. B., Morris, G. M., Rudd, P. M., Dwek, R. A., Stanfield, R. L., Burton, D. R. & other authors (2001). Crystal structure of a neutralizing human IGG against HIV-1: a template for vaccine design. Science 293, 1155-1159. Walker, L. M., Bowley, D. R. & Burton, D. R. (2009). Efficient recovery of high-affinity antibodies from a single-chain Fab yeast display library. J Mol Biol 389, 365-375. Wu, X., Yang, Z. Y., Li, Y., Hogerkorp, C. M., Schief, W. R., Seaman, M. S., Zhou, T., Schmidt, S. D., Wu, L. & other authors (2010). Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329, 856-861. Zhou, T., Xu, L., Dey, B., Hessell, A. J., Van Ryk, D., Xiang, S., Yang, X., Zhang, M., Zwick, M. B.& other authors (2007). Structural definition of a conserved neutralization epitope on HIV-1 gp120. Nature 445, 732–737.

298 299 300

Zhou, T., Georgiev, I., Wu, X., Yang, Z. Y., Dai, K., Finzi, A., Kwon, Y. D., Scheid, J. F., Shi, W. & other authors (2010). Structural basis for broad and potent neutralization of HIV-1 by antibody VRC01. Science 329, 811-817.

301 302 303 304 305 306

307 308

Figure legends

309

Figure 1

310 311

Sequences of the BNMAbs-selected EMs are compared to the parental HXB2 env gene, with the nucleotide number in italics to the left. EM3 is the HJ16/A12/b12 triply selected virus.

312

Figure 2

313

316

HXB2 (white discs), SDM(A12), SDM(b12) and HJ16-resistant pseudotyped HIV-1 virions (black discs) were assessed for neutralization resistance against against (a) A12, b12 and HJ16 and (b) VRC01, J3 and CD4-IgG2. The % of neutralization is shown on the y-axis in the presence of BNMAbs (in bold at the bottom of the graph) at different concentrations (x-axis: ng x ml-1).

317

Figure 3

318 319 320 321

A model of the crystal structure of the HIV-1 Env trimer, where identified AA changes are indicated with arrows. HXB2 gp120 is adapted from 3JWD (Pancera et al., 2010). The trimer is adapted from 4NCO.

314 315

322 323 324 325

Figure Click here to download Figure: Dreja 10 Feb2015.pptx

Figure 1

HXB2

360 FKQS SGGDPE IV THSFNCGG E FFYCNSTQL FNSTWFNST W STEGSNNT EG SD TITLPCRI KQIIN MWQKV GKA MYAPPIS

GQ IRCSSNIT G LLLTRDGGN SNNESEIFR P

EM(b12)

---- -E---- -- -------- - --------- --------- - -------- -- -- -------- ----- ----- --- ------- -- -------- - --------- --------- -

EM(A12)

---- ------ -- --N----- - --------- --------- - -------- -- -- -------- ----- ----- --- ------- -- -------- - --------- --------- -

EM(HJ16F) ---- ------ -- -------- - --------- --------- - -------- -- -- -------- ----- ----- --- ------- -- -------- - --------- ----F---- EM(HJ16P) ---- ------ -- -------- - --------- --------- - -------- -- -- -------- ----- ----- --- ------- -- -------- - --------- ----P---- EM(HJ16Y) ---- ------ -- -------- - --------- --------- - -------- -- -- -------- ----- ----- --- ------- -- -------- - --------- ----Y---- EM(HJ16S) ---- ------ -- -------- - --------- --------- - -------- -- -- -------- ----- ----- --- ------- -- -------- - --------- --S------ EM(HJ16D) ---- ------ -- -------- - --------- --------- - -------- -- -- -------- ----- ----- --- ------- -- -------- - -------D- --------- -

EM(HJ16R) ---- ------ -- -------- - --------- --------- - -------- R- -- -------- ----- ----- --- ------- -- -------- - --------- --------- EM3

---- -E---- -- --N----- - --------- --------- - -------- -- -- -------- ----- ----- --- ------- -- -------- - --------- --S------ -

psHJ16 (S465Y)

psHJ16 (N463S)

psHJ16 (G459D)

psHJ16 (E409R)

% neutralization % neutralization % neutralization

psHJ16 (S465P)

% neutralization

psHJ16F (S465F)

% neutralization

SDM(b12) G366E

% neutralization

WT HXB2

% neutralization

SDM(A12) S375N

% neutralization

Figure 2A 120 100 80 60 40 20 0 120 100 80 60 40 20 0 120 100 80 60 40 20 0 120 100 80 60 40 20 0 120 100 80 60 40 20 0 120 100 80 60 40 20 0 120 100 80 60 40 20 0 -20 120 100 80 60 40 20 0 -20

A12

1

10

1

10

1

1

1

1

1

1

b12 120 100 80 60 40 20 0 120 100 1000 100 80 60 40 20 0 100 1000 120

100 80 60 40 20 0 120 10 100 1000 100 80 60 40 20 0 120 10 100 1000 100 80 60 40 20 0 10 100 1000120 100 80 60 40 20 0 10 100 1000 120 100 80 60 40 20 0 10 100 1000120 100 80 60 40 20 ng x ml-1 0 10

100

1000

1

10

1

10

1

10

1

10

1

10

1

10

1

120 100 80 60 40 20 0 100 1000 120 100 80 60 40 20 0 100 1000 120 100 80 60 40 20 0 120 100 1000 100 80 60 40 20 0 100 1000 120 100 80 60 40 20 0 100 1000 120 100 80 60 40 20 0 -20 100 1000 120

10 100 1000

ng x ml-1 1

10

HJ16

100 1000

100 80 60 40 20 0 -20 120 100 80 60 40 20 0 -20

1

10 100 1000

1

10 100 1000

1

10 100 1000

1

10

100 1000

1

10

100 1000

1

10

100 1000

1

10

100 1000

ng x ml-1 1

10

100

1000

psHJ16 (S465Y)

psHJ16 (N463S)

psHJ16 (G459D)

psHJ16 (E409R)

120 100 80 60 40 20 0 120 1 10 100 1000 100 80 60 40 20 0 120 -20 1 10 100 1000 100 80 60 40 20 0120 -20100 1 10 100 1000 80 60 40 20 0 -20 1 10 100 1000 120 100 80 60 40 20 0 -20 1 10 100 1000 120 100 80 60 40 20 0 -20 1 10 100 1000 120 100 80 60 40 20 ng x ml-1 0 1 10 100 1000 -20

J3

120 100 80 60 40 20 0

120 100 80 60 40 20 0 120 100 80 60 40 20 0 120 100 80 60 40 20 0

120 100 80 60 40 20 0

120 100 80 60 40 20 0

120 100 80 60 40 20 0

120 100 80 60 40 20 0

120 100 80 60 40 20 0 120 100 80 60 40 20 0

120 100 80 60 40 20 0

% neutralization

% neutralization

120 100 80 60 40 20 0 120 100 80 60 40 20 0

% neutralization

psHJ16 (S465P)

VRC01

% neutralization

psHJ16F (S465F)

120 100 80 60 40 20 0

% neutralization

SDM(b12) G366E

% neutralization.

WT HXB2

% neutralization

SDM(A12) S375N

% neutralization

Figure 2B

1

10

120 100 80 60 40 20 0

100 1000

ng x ml-1 1

10

100 1000

CD4-IgG2

1

10 100 1000

1

10 100 1000

120 100 80 60 40 20 120 100 80 60 40 20 0 -20

ng x ml-1 1

10

100 1000

Figure 3

HJ16: N463S HJ16: G459D HJ16: E409R

A12: S375N

b12: G366E

gp41

HJ16: N463S HJ16: G459D HJ16: E409R b12: G366E

Glycan gp120s

CD4BS A12: S375N

CD4 binding site broadly neutralizing antibody selection of HIV-1 escape mutants.

All human immunodeficiency virus type-1 (HIV-1) viruses use CD4 to enter cells. Consequently, the viral envelope CD4-binding site (CD4bs) is relativel...
1MB Sizes 0 Downloads 13 Views